WO2004062587A2 - Therapie d'administration orale d'insuline le soir - Google Patents

Therapie d'administration orale d'insuline le soir Download PDF

Info

Publication number
WO2004062587A2
WO2004062587A2 PCT/US2004/000273 US2004000273W WO2004062587A2 WO 2004062587 A2 WO2004062587 A2 WO 2004062587A2 US 2004000273 W US2004000273 W US 2004000273W WO 2004062587 A2 WO2004062587 A2 WO 2004062587A2
Authority
WO
WIPO (PCT)
Prior art keywords
insulin
oral administration
levels
hours
oral
Prior art date
Application number
PCT/US2004/000273
Other languages
English (en)
Other versions
WO2004062587A3 (fr
Inventor
Michael Goldberg
Ehud Arbit
Original Assignee
Emisphere Technologies, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Emisphere Technologies, Inc. filed Critical Emisphere Technologies, Inc.
Priority to NZ541058A priority Critical patent/NZ541058A/en
Priority to CA2511530A priority patent/CA2511530C/fr
Priority to US10/541,433 priority patent/US7384914B2/en
Priority to JP2006500813A priority patent/JP5452843B2/ja
Priority to ES04700388.4T priority patent/ES2465496T3/es
Priority to AU2004204727A priority patent/AU2004204727B8/en
Priority to EP04700388.4A priority patent/EP1592438B1/fr
Publication of WO2004062587A2 publication Critical patent/WO2004062587A2/fr
Publication of WO2004062587A3 publication Critical patent/WO2004062587A3/fr
Priority to HK06105431.0A priority patent/HK1085133A1/xx

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/28Insulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • This invention relates to the oral delivery of insulin in a therapeutically effective amount to the bloodstream as part of a therapeutic regimen for the treatment of diabetes.
  • This invention further relates to compositions of a delivery agent and insulin for oral administration that facilitates insulin transport in a therapeutically effective amount to the bloodstream for the treatment of diabetes.
  • This invention further relates to the oral administration of dosage forms of insulin and a delivery agent at or shortly before bedtime for the treatment of diabetes.
  • biological macromolecules namely biological polymers such as proteins and polypeptides
  • proteins are increasingly being use in many diverse areas of science and technology.
  • proteins are employed as active agents in the fields of pharmaceuticals, vaccines and veterinary products.
  • biological macromolecules as active agents in pharmaceutical compositions is often severely limited by the presence of natural barriers of passage to the location where the active agent is required. Such barriers include the skin, lipid bi-layers, mucosal membranes, severe pH conditions and digestive enzymes.
  • biological macromolecules are large and are amphipathic in nature. More importantly, the active conformation of many biological macromolecules may be sensitive to a variety of environmental factors, such as temperature, oxidizing agents, pH, freezing, shaking and shear stress. In planning oral delivery systems comprising biological macromolecules as an active agent for drug development, these complex structural and stability factors must be considered.
  • delivery vehicles can be used to facilitate absorption through the gastro-intestinal tract. These delivery vehicles must be able to release active molecules, at a rate that is consistent with the needs of the particular patient or the disease process.
  • insulin contributes to the normal regulation of blood glucose levels through its release by the pancreas, more specifically by the ⁇ -cells of a major type of pancreatic tissue (the islets of Langerhans), so that the glucose can be used as a source of energy.
  • Insulin secretion is a regulated process that, in normal subjects, provides stable concentrations of glucose in blood during both fasting and feeding.
  • insulin is secreted from the pancreas into the portal vein, which carries the insulin to the liver.
  • the liver utilizes and/or metabolizes a large portion of the insulin that it receives from the portal circulation.
  • Blood glucose concentration is the principal stimulus to insulin secretion in healthy humans.
  • Glucose enters the pancreatic 3-cell by facilitated transport and is then phosphorylated by glucokinase.
  • Expression of glucokinase is primarily limited to cells and tissues involved in the regulation of glucose metabolism, such as the liver and the pancreatic /..-cells.
  • the capacity of sugars to undergo phosphorylation and subsequent glycolysis correlates closely with their ability to stimulate insulin release. It is noted that not all tissues are dependent on insulin for glucose uptake. For example, the brain, kidneys and red blood cells are insulin independent tissues, while the liver, adipose and muscle are insulin dependent tissues.
  • Diabetes Mellitus (“diabetes”) is a disease state in which the pancreas does not release insulin at levels capable of controlling glucose levels and/or in which muscle, fat and liver cells respond poorly to no ⁇ nal insulin levels because of insulin resistance. Diabetes Mellitus is classified into two types: Type 1 and Type 2. Approximately 5 to 10% of diagnosed cases of diabetes are attributed to Type 1 diabetes, and approximately 90% to 95% to type 2 diabetes.
  • Type 1 diabetes is diabetes that is insulin dependent and usually first appears in young people.
  • the islet cells of the pancreas stop producing insulin mainly due to autoimmune destruction and the patient must inject himself with the missing hormone.
  • Type 2 diabetes is non-insulin dependent diabetes, which may be caused by a combination of insulin resistance (or decreased insulin sensitivity) and, in later stages, insufficient insulin secretion. This is the most common type of diabetes in the Western world. Close to 8% of the adult population of various countries around the world, including the United States, have Type 2 diabetes, and about 30% of these patients will need to use exogenous insulin at some point during their life spans due to secondary pancreas exhaustion and the eventual cessation of insulin production.
  • Insulin resistance is also prevalent in the population, especially in overweight individuals, in those with risk of diabetes (i.e., pre-diabetic, wherein blood glucose levels are higher than normal but not yet high enough to be diagnosed as diabetes) and in individuals with type 2 diabetes who produce enough insulin but whose tissues have a diminished ability to adequately respond to the action of insulin.
  • diabetes i.e., pre-diabetic, wherein blood glucose levels are higher than normal but not yet high enough to be diagnosed as diabetes
  • type 2 diabetes who produce enough insulin but whose tissues have a diminished ability to adequately respond to the action of insulin.
  • the pancreatic ⁇ -cells initially increase their insulin production such that insulin resistant individuals have high plasma insulin levels.
  • the cells start to decompensate and exhaust, and insulin secretion is reduced at later stages of diabetes.
  • Diabetes is the sixth leading cause of death in the United States and accounted for more than 193,000 deaths in 1997. However, this is an underestimate because complications resulting from diabetes are a major cause of morbidity in the population. Diabetes is associated with considerable morbidity and mortality in the form of cardiovascular disease, stroke, digestive diseases, infection, metabolic complications, ophthalmic disorders, neuropathy, kidney disease and failure, peripheral vascular disease, ulcerations and amputations, oral complications, and depression. Thus, diabetes contributes to substantially many deaths that are ultimately ascribed to other causes.
  • the main cause of mortality with Diabetes Mellitus is long term micro- and macro- vascular disease.
  • Cardiovascular disease is responsible for up to 80% of the deaths of type 2 diabetic patients, and diabetics have a two- to four-fold increase in the risk of coronary artery disease, equal that of patients who have survived a stroke or myocardial infarction.
  • heart disease, high blood pressure, heart attacks and strokes occur two to four times more frequently in adult diabetics than in adult non-diabetics.
  • This increased risk of coronary artery disease combined with an increase in hypertensive cardiomyopathy manifests itself in an increase in the risk of congestive heart failure.
  • These vascular complications lead to neuropathies, retinopathies and peripheral vascular disease.
  • Diabetic retinopathy (the breakdown of the lining at the back of the eye) is the leading cause of blindness in adults aged 20 through 74 years, and diabetic kidney disease, e.g., nephropathy (the inability of the kidney to properly filter body toxins), accounts for 40%) of all new cases of end-stage renal disease (kidney failure). Furthermore, diabetes is also the leading cause for amputation of limbs in the United States. Diabetes causes special problems during pregnancy, and the rate of congenital malformations can be five times higher in the children of women with diabetes. [0013] Insulin resistance plays an important role in the pathogenesis of hyperglycemia in type 2 diabetes, which eventually induces the development of diabetic complications. Furthermore, insulin resistance ostensibly plays a role in the pathogenesis of macro vascular disease, cardiovascular diseases and microvascular disease.
  • insulin therapy is essential and is intended to replace the absent endogenous insulin with an exogenous insulin supply.
  • therapy has consisted of oral antidiabetic agents, which increase insulin sensitivity and/or insulin secretion, and insulin if, and when, the oral agents fail.
  • Hyperinsulinemia can occur by the administration of insulin in a location (and manner) that is not consistent with the normal physiological route of delivery. Insulin circulates in blood as the free monomer, and its volume of distribution approximates the volume of extracellular fluid. Under fasting conditions, the concentration of insulin in portal blood is, e.g., about 2-4 ng/mL, whereas the systemic (peripheral) concentration of insulin is, e.g., about 0.5 ng/mL, in normal healthy humans, translating into, e.g., a 5:1 ratio. In human diabetics who receive insulin via subcutaneous injection, the ratio is changed to about 0.75:1.
  • the liver does not receive the necessary concentrations of insulin to adequately control blood glucose.
  • Elevated systemic levels of insulin may lead to increased glucose uptake, glycogen synthesis, glycolysis, fatty acid synthesis, cortisol synthesis and triacylglycerol synthesis, leading to the expression of key genes that result in greater utilization of glucose.
  • Insulin absorbed in the gastrointestinal tract mimics the physiology of insulin secreted by the pancreas because both are released into the portal vein and carried directly to the liver before being delivered into the peripheral circulation. Absorption into the portal circulation maintains a peripheral-portal insulin gradient that regulates insulin secretion. In its first passage through the liver, roughly 60% of the insulin is retained and metabolized, thereby reducing the incidence of peripheral hyperinsulinemia, a factor linked to complications in diabetes. A not uncommon, and serious, complication of insulin treatment and other oral antidiabetic agents is hypoglycemia.
  • insulin exemplifies the problems confronted in the art in designing an effective oral drug delivery system for biological macromolecules. Insulin absorption in the gastrointestinal tract is prevented presumably by its molecular size and its susceptibility for enzymatic degradation. The physicochemical properties of insulin and its susceptibility to enzymatic digestion have precluded the design of a commercially viable oral or alternate delivery system.
  • Emisphere Technologies, Inc. has developed compositions of insulin that are orally administrable, e.g., absorbed from the gastrointestinal tract in adequate concentrations, such that the insulin is bioavailable and bioactive following oral administration and provide sufficient absorption and pharmacokinetic/phannacodynamic properties to provide the desired therapeutic effect, i.e., cause a reduction of blood glucose, as disclosed in U.S. Patent Applications Nos. 10/237,138, 60/346,746, 60/347,312, 60/368,617, 60/374,979, 60/389,364, 60/438,195, 60/438,451, 60/438,444, 60/452,660 and 60/488,465, as well as in International Patent Application Publications Nos. WO 03/057170, WO 03/057650 and WO02/02509, all assigned to Emisphere Technologies, Inc., all of which are incorporated herein by reference.
  • the novel drug delivery technology of Emisphere Technologies, Inc. is based upon the design and synthesis of low molecular weight compounds called "delivery agents.”
  • the delivery agent which is in a preferred embodiment 4-CNAB (sodium N-[4-(4-chloro-2 hydroxybenzoyl)amino]butyrate) enables the gastrointestinal absorption of insulin. It is believed that the mechanism of this process is that 4-CNAB interacts with insulin non-covalently, creating more favorable physical-chemical properties for absorption. Once across the gastrointestinal wall, insulin disassociates rapidly from 4-CNAB and reverts to its normal, pharmacologically active state. 4-CNAB is not intended to possess any inherent pharmacological activity and serves only to increase the oral bioavailability of insulin by facilitating the transport of insulin across the gastrointestinal wall. The pharmacology of insulin is the desired therapeutic effect and is well characterized.
  • the oral dosing method developed by Emisphere Technologies, Inc. mimics natural physiology, namely, the ratio of concentration of unmodified insulin in the portal circulation to that in the systemic circulation approaches the normal physiological ratio, e.g., from about 2:1 to about 6:1.
  • the effect of this route of dosing is two fold.
  • a greater control of glucose may be achieved.
  • Various studies have shown that intraportal delivery of insulin can yield a comparable control of glucose at infusion rates lower than those required by peripheral administration. Because the insulin will undergo substantial ( ⁇ 50%) first-pass metabolism prior to entering the systemic circulation, a lower serum concentration and total exposure is achieved. This may, in turn, alleviate any detrimental effects of insulin on non-target tissues.
  • Insulin/4-CNAB capsules were evaluated by Emisphere Technologies, Inc. in a nonclinical program that included pharmacological screening, pharmacokinetic and metabolic profiles, and toxicity assessments in rats and monkeys. These studies in rats and monkeys showed that 4-CNAB is absorbed rapidly following oral administration.
  • Insulin/4-CNAB capsules were also evaluated by Emisphere Technologies, Inc. in clinical studies for the safety, pharmacokinetics, pharmacodynamics, and the effect of food on the absorption of insulin.
  • 4-CNAB was shown to enhance the gastrointestinal absorption of insulin following oral administration in diabetic patients and healthy subjects.
  • Oral administration of Insulin/4-CNAB capsules resulted in rapid absorption of both insulin and 4-CNAB, and the insulin absorbed orally in combination with 4-CNAB was pharmacologically active, as demonstrated by a reduction of blood glucose in healthy and diabetic subjects and by a blunting of postprandial glucose excursion in diabetic patients.
  • These studies suggest that oral administration of a fonnulation of insulin/4-CNAB is well-tolerated and reduces blood glucose concentrations in both healthy subjects and diabetic patients.
  • first-phase insulin secretion takes place 5 to 20 minutes after the start of a meal, and this effect has a well-known impact on prandial glucose homeostasis.
  • the loss of first-phase insulin secretion is a characteristic feature of Type 2 diabetic patients in the early stages of the disease, and it is also observed in prediabetic states, namely individuals with impaired glucose tolerance.
  • the stimulatory effect of glucagon on gluconeogenesis is not suppressed and may contribute to the development of prandial hyperglycemia.
  • plasma glucose concentrations are correlated with hepatic glucose output. Therefore, restoration of first-phase insulin secretion at the time of meal ingestion should suppress prandial hepatic glucose output and subsequently improve the blood glucose profile.
  • certain short acting insulin formulations because of the speed with which the insulin provides a blood glucose lowering effect, may, between the time of administration of insulin and the time of ingestion of the meal, contribute to a lowering of blood glucose to a level that could range from subclinical hypoglycemia to more undesirable effects.
  • compositions comprising insulin and a delivery agent for oral administration at or shortly prior to bedtime.
  • compositions comprising insulin and a delivery agent for oral administration at or shortly prior to bedtime.
  • a delivery agent that facilitates insulin transport in a therapeutically effective amount to the bloodstream for the treatment of diabetes, for the treatment of impaired glucose tolerance, for the purpose of achieving glucose homeostasis, for the treatment of early stage diabetes, for the treatment of late stage diabetes, and/or to serve as replacement therapy for type I diabetic patients.
  • It is a further object of the invention to provide methods for prophylactically sparing pancreatic beta cell function and for preventing beta cell death or dysfunction in a mammal that has impaired glucose tolerance or early stage diabetes.
  • the invention is directed in part to a method of treatment of diabetes in mammals, comprising orally administering one or more unit doses of the dosage forms described above and in further sections of the present specification at or shortly prior to bedtime.
  • Mammals includes but is not limited to rodents, aquatic mammals, domestic animals such as dogs and cats, farm animals such as sheep, pigs, cows and horses, and humans.
  • the mammal is a human.
  • the invention is further directed in part to a method of treatment of impaired glucose tolerance, early stage diabetes and late stage diabetes in mammals, and of achieving glucose homeostasis, comprising administering one or more unit doses of the dosage forms described in the present specification at or shortly prior to bedtime.
  • the oral insulin formulation is administered to such patients on a chronic basis, e.g., for at least about 2 weeks, and can be administered for the life of the patient.
  • the present invention is also directed in part to a method of treating diabetes and reducing the incidence of systemic hyperinsulinemia associated with chronic dosing of insulin, comprising orally administering to a diabetic patient at or shortly prior to bedtime a dose of insulin and a delivery agent that facilitates the absorption of the dose of insulin from the gastrointestinal tract to provide a therapeutically effective reduction and/or control in blood glucose and a mean systemic blood insulin concentration of the diabetic patient that is reduced relative to the mean systemic blood insulin concentration provided by subcutaneous injection of insulin in an amount effective to achieve equivalent reduction and/or control in a population of diabetic patients.
  • the oral insulin formulation is administered to such patients on a chronic basis, e.g., for at least about 2 weeks, and can be administered for the life of the patient.
  • the mean values of insulin concentration determination obtained in patients who have been administered subcutaneous insulin are well known to those skilled in the art.
  • the present invention is further directed to a method for prophylactically sparing beta cell function in a mammal which has impaired glucose tolerance or early stage diabetes mellitus, comprising administering an orally effective dose of a pharmaceutical formulation comprising insulin (as described herein) at nighttime, e.g., at or shortly prior to bedtime.
  • the oral insulin formulation is administered to such patients on a chronic basis, e.g., for at least about 2 weeks, and can be administered for the life of the patient.
  • the present invention is further directed to a method for preventing beta cell death or dysfunction in a mammal which has impaired glucose tolerance or early stage diabetes mellitus, comprising administering an orally effective dose of a pharmaceutical formulation comprising insulin at nighttime.
  • a pharmaceutical formulation comprising insulin at nighttime.
  • the oral insulin formulation is administered to such patients at or shortly prior to bedtime on a chronic basis, e.g., for at least about 2 weeks.
  • the present invention is further directed to a method for long-term protection of a mammal which has impaired glucose tolerance or early stage diabetes mellitus from developing overt or insulin dependent diabetes, comprising administering an orally effective dose of a pharmaceutical formulation comprising insulin at nighttime.
  • a pharmaceutical formulation comprising insulin at nighttime.
  • the oral insulin formulation is administered to such patients at or shortly prior to bedtime on a chronic basis, e.g., for at least about 2 weeks.
  • the present invention is further directed to a method for delaying the onset of overt or insulin dependent diabetes in a mammal which has impaired glucose tolerance or early stage diabetes mellitus, comprising administering an orally effective dose of a pharmaceutical formulation comprising insulin at nighttime.
  • a pharmaceutical formulation comprising insulin at nighttime.
  • the oral insulin formulation is administered to such patients at or shortly prior to bedtime on a chronic basis, e.g., for at least about 2 weeks.
  • the invention is further directed to a method of providing a therapeutically effective orally administrable unit dose of unmodified insulin, comprising combining from about 2 to about 23 mg of unmodified insulin with from about 100 to about 600 mg of a pharmaceutically acceptable delivery agent which facilitates absorption of said insulin from the gastrointestinal tract, and orally administering said unit dose to a diabetic patient at or shortly prior to bedtime to provide a therapeutic effect.
  • the total weight of the unit dose is from about 102 mg to about 800 mg.
  • the method comprises the following steps: contacting insulin with said delivery agent, and thereafter orally administering the pharmaceutical composition to the patient at or shortly prior to bedtime.
  • the method comprises administering the insulin and the delivery agent in such a manner that the insulin and delivery agent contact each other in-vivo (e.g., in the stomach), such that the delivery agent is available to facilitate absorption of the insulin through the gastrointestinal mucosa and into the portal vein, the normal physiologic route.
  • the phrase administered "at nighttime” or “at or shortly before bedtime” means administered less than about 3 hours, preferably les than about 2 hours and more preferably less than about 1 hour prior to a prolonged period of sleep, or relative physical and/or mental inactivity, and fast, e.g., overnight. Whereas overnight typically means from the late night (p.m.) hours to the early morning (a.m.) hours, it could mean any period of a sleep-wake cycle during which a person obtains his/her necessary period of sleep.
  • administration should also occur at least about one hour, preferably at least about 1.5 hours, more preferably at least about 2 hours and still more preferably at least about 2 to about 3 hours after the last meal of the day.
  • the invention is directed in part to an oral solid dosage form comprising a dose of unmodified insulin that, when administered at or shortly before bedtime, achieves a maximum control of post prandial blood glucose concentration in diabetic patients within one hour post-administration.
  • the invention is directed in part to an oral solid dosage form comprising a dose of unmodified insulin that achieves a reduction in blood glucose concentration in diabetic patients comparable to a subcutaneous insulin injection in those patients, while providing a lower (e.g., 20%> or greater) totals dose of insulin in the peripheral blood circulation under acute, sub-acute and chronic conditions as compared to the peripheral blood insulin concentration obtained via the subcutaneous injection.
  • the invention is also directed in part to an oral solid dosage form comprising a dose of unmodified insulin that achieves a therapeutically effective reduction in blood glucose after oral administration to a diabetic patient, and which maintains a physiological (portal/ peripheral) gradient, and in certain embodiments provides a ratio of portal vein insulin concentration to peripheral blood insulin concentration from about 2.5: 1 to about 6:1, and preferably from about 4:1 to about 5:1.
  • the invention is further directed in part to an oral dosage form comprising a dose of unmodified insulin that achieves a therapeutically effective reduction in blood glucose after oral administration to diabetic patients, the oral solid dosage form providing an insulin t max at a time point from about 0.25 to about 1.5 hours after oral administration to said patients, at least about 80% of the blood glucose concentration reduction caused by said dose of insulin occurring within about 2 hours after oral administration of said dosage form.
  • the oral dosage form is solid, such as a gelatin capsule or a tablet.
  • the dose of unmodified insulin contained in the dosage form is from about 50 Units to about 600 Units (from about 2 to about 23 mg), preferably from about 100 Units (3.8 mg) to about 450 Units (15.3 mg) insulin, more preferably from about 200 Units (7.66 mg) to about 350 Units (13.4 mg), and still more preferably about 300 Units (11.5 mg), based on the accepted conversion of factor of 26.11 Units per mg.
  • the dosage forms of the invention provide a t max for insulin at about 0.1 to about 1.5 hours, and more preferably by about 0.25 to about 0.5 hours, after oral administration.
  • the t raax for insulin occurs at less than about 100 minutes after oral administration of the composition, preferably at less than about 45 minutes, more preferably at less than about 40 minutes, and still more preferably at about 22 minutes after oral administration of the composition.
  • the composition provides a t max for maximum control of glucose excursion at about 0.25 to about 1.5 hours, more preferably at about 0J5 to about 1.25 hours, after oral administration.
  • the t max for glucose control occurs preferably at less than about 120 minutes, more preferably at less than about 80 minutes, and still more preferably at about 45 minutes to about 60 minutes, after oral administration of the composition.
  • the dosage forms begin delivering insulin into the portal circulation (via absorption through the mucosa of the stomach) to achieve peak levels within about 30 minutes or less.
  • the dose of unmodified insulin in the absence of a delivery agent, is not adequately absorbed from the gastrointestinal tract when administered orally to render a desired effect.
  • the dose of insulin in the absence of a delivery agent, is not sufficiently absorbed when orally administered to a patient to provide a desirable therapeutic effect but said dose provides a desirable therapeutic effect when administered to said patient by another route of administration.
  • the invention in such embodiments is further directed to an oral dosage form comprising a dose of unmodified insulin together with a pharmaceutically acceptable delivery agent in an amount effective to facilitate the absorption of said insulin, such that a therapeutically effective amount of said dose of insulin is absorbed from the gastrointestinal tract of diabetic patients.
  • the pharmaceutical composition comprises from about 1 mg to about 800 mg of said delivery agent, preferably about 50 to about 600, more preferably from about 100 to about 400, most preferably about 200.
  • the composition provides a peak plasma delivery agent concentration C max from about 1,000 and about 150,000 ng/ml, and a t max at about 0.25 to about 1.5 hours, and more preferably by about 0.25 to about 0J5 hours, most preferably 0.5 hours, after oral administration.
  • a preferred delivery agent is identified via chemical nomenclature as 4-[(4-chloro, 2-hydroxybenzoyl)amino]butanoic acid.
  • the delivery agent is a sodium salt, preferably monosodium salt.
  • the same compound is identified by the alternative nomenclature monosodium N-(4-chlorosalicyloyl)- 4-aminobutyrate, or by the short name "4-CNAB”.
  • Diabetic patient « refers to a mammal with a form of diabetes.
  • IGT means impaired glucose tolerance
  • Diabetes or Diabetes Mellitus ⁇ is deemed to encompass type 1 and type 2 diabetes mellitus, unless specifically specified otherwise.
  • Delivery agent refers to carrier compounds or carrier molecules that are effective in the oral delivery of therapeutic agents, and may be used interchangeably with “carrier”.
  • Therapeutically effective amount of insulin ⁇ refers to an amount of insulin included in the dosage forms of the invention which is sufficient to achieve a clinically relevant control of blood glucose concentrations in a human diabetic patient either in the fasting state or in the fed state effective, during the dosing interval.
  • Effective amount of delivery agent an amount of the delivery agent that promotes the absorption of a therapeutically effective amount of the drug from the gastrointestinal tract.
  • Organic solvents refers to any solvent of non-aqueous origin, including liquid polymers and mixtures thereof.
  • Organic solvents suitable for the present invention include: acetone, methyl alcohol, methyl isobutyl ketone, chloroform, 1-propanol, isopropanol, 2-propanol, acetonitrile, 1- butanol, 2-butanol, ethyl alcohol, cyclohexane, dioxane, ethyl acetate, dimethylformamide, dichloroethane, hexane, isooctane, methylene chloride, tert-butyl alchohol, toluene, carbon tetrachloride, or combinations thereof.
  • Peptide refers to a polypeptide of small to intermediate molecular weight, usually 2 or more amino acid residues and frequently but not necessarily representing a fragment of a larger protein.
  • Protein ⁇ refers to a complex high polymer containing carbon, hydrogen, oxygen, nitrogen and usually sulfur and composed of chains of amino acids connected by peptide linkages. Proteins in this application refer to glycoproteins, antibodies, non-enzyme proteins, enzymes, hormones and sub-units of proteins, such as peptides. The molecular weight range for proteins includes peptides of 1000 Daltons to glycoproteins of 600 to 1000 kiloDaltons.
  • Reconstitution - refers to dissolution of compositions or compositions in an appropriate buffer or pharmaceutical composition.
  • Unit-Dose Forms refers to physically discrete units suitable for human and animal subjects and packaged individually as is known in the art. It is contemplated for purposes of the present invention that dosage forms of the present invention comprising therapeutically effective amounts of insulin may include one or more unit doses (e.g., tablets, capsules, powders, semisolids (e.g. gelcaps or films), liquids for oral admisistration, ampoules or vials for injection, loaded syringes) to achieve the therapeutic effect. It is further contemplated for the purposes of the present invention one preferred embodiment of the dosage form is an oral dosage form.
  • unit doses e.g., tablets, capsules, powders, semisolids (e.g. gelcaps or films), liquids for oral admisistration, ampoules or vials for injection, loaded syringes
  • Unmodified insulin refers to insulin prepared in any pharmaceutically acceptable manner or from any pharmaceutically acceptable source which is not conjugated with an oligomer such as that described in U.S. Patent No. 6,309,633 and/or which not has been subjected to amphiphilic modification such as that described in U.S. Patent Nos. 5,359,030; 5,438,040; and/or 5,681,811, which patents are hereby incorporated by reference in their entireties.
  • mean when preceding a phannacokinetic value (e.g., mean t max ), represents the arithmetic mean value of the pharmacokinetic value unless otherwise specified.
  • mean baseline level means the measurement, calculation or level of a certain value that is used as a basis for comparison, which is the mean value over a statistically significant number of subjects, e.g., across a single clinical study or a combination of more than one clinical study.
  • C max is the highest plasma concentration of the drug observed within the dosing interval.
  • t max is the time post-dose at which C max is observed.
  • AUC area under the plasma concentration-time curve, as calculated by the trapezoidal rule over the complete sample collection interval.
  • multiple dose means that the human patient has received at least two doses of the drug composition in accordance with the dosing interval for that composition.
  • single dose means that the human patient has received a single dose of the drug composition and the drug plasma concentration has not achieved steady state. Unless specifically designated as “single dose” or at “steady-state”, the pharmacokinetic parameters disclosed and claimed herein encompass both single dose and steady-state conditions.
  • Bioavailability means the degree or ratio (%>) to which a drug or agent is absorbed or otherwise available to the treatment site in the body. This is calculated by the formula
  • Biopotency means the degree or ratio (%) to which a drug or agent is effective to the treatment site in the body. This is calculated by the formula
  • Figure 1 is a table of data (blood glucose, insulin and C-peptide) collected in the morning after nighttime dosing of insulin and 4-CNAB for each subject compared to that subject's own baseline levels.
  • Figure 2 is a table of data (insulin and C-peptide) collected at night prior to nighttime dosing of insulin and 4-CNAB for each subject.
  • Figure 3 is a bar graph showing the effect of nighttime dosing of insulin and 4-CNAB on blood glucose concentration.
  • Figure 4 is a bar graph showing the effect of nighttime dosing of insulin and 4-CNAB on blood C-peptide concentration.
  • Figure 5 is a bar graph showing the effect of nighttime dosing of insulin and 4-CNAB on blood insulin concentration.
  • the invention further provides a method of treatment of diabetes, impaired glucose tolerance, early stage diabetes, and late stage diabetes in animals, preferably humans, and a method of achieving glucose homeostasis, comprising administering one or more unit doses of the dosage forms described in the present specification at or shortly before bedtime, preferably on a chronic basis.
  • the invention further provides a method for treating animals, preferably humans with an oral dosage form of a pharmaceutical composition that includes: insulin or a pharmaceutically acceptable salt thereof and an effective amount of a delivery agent or a pharmaceutically acceptable salt thereof that renders insulin orally absorbed (i.e., bioavailable), by administering said composition to said animals, preferably humans on a chronic basis at or shortly before bedtime.
  • a pharmaceutical composition that includes: insulin or a pharmaceutically acceptable salt thereof and an effective amount of a delivery agent or a pharmaceutically acceptable salt thereof that renders insulin orally absorbed (i.e., bioavailable)
  • the invention further provides a method of treating diabetes and/or reducing the incidence of systemic hyperinsulinemia associated with chronic dosing of insulin, comprising orally administering to a diabetic patient at or shortly before bedtime, preferably on a chronic basis, a dose of insulin and a delivery agent that facilitates the absorption of the dose of insulin from the gastrointestinal tract.
  • the present invention is further directed to method for prophylactically sparing beta cell function in a mammal which has impaired glucose tolerance or early stage diabetes mellitus, comprising administering an orally effective dose of a pharmaceutical formulation comprising insulin (as described herein) at or shortly prior to bedtime on a chronic basis, e.g., for at least about 2 weeks, or for the life of the patient.
  • the present invention is further directed to a method for preventing beta cell death or dysfunction in a mammal which has impaired glucose tolerance or early stage diabetes mellitus, comprising administering an orally effective dose of a pharmaceutical formulation comprising insulin at or shortly prior to bedtime on a chronic basis, e.g., for at least about 2 weeks.
  • the present invention is further directed to a method for long-term protection of a mammal which has impaired glucose tolerance or early stage diabetes mellitus from developing overt or insulin dependent diabetes, comprising administering an orally effective dose of a pharmaceutical formulation comprising insulin at or shortly prior to bedtime on a chronic basis, e.g., for at least about 2 weeks.
  • the present invention is further directed to a method for delaying the onset of overt or insulin dependent diabetes in a mammal which has impaired glucose tolerance or early stage diabetes mellitus, comprising administering an orally effective dose of a pharmaceutical formulation comprising insulin at or shortly prior to bedtime on a chronic basis, e.g., for at least about 2 weeks.
  • the present invention provides a method of administering insulin and pharmaceutical compositions useful for administering insulin such that the insulin is bioavailable and biopotent.
  • the delivery agent enables insulin to be orally absorbable through the mucosa of the stomach and facilitates the absorption of insulin administered therewith (either in the same dosage form, or simultaneously therewith), or sequentially (in either order, as long as both the delivery agent and insulin are administered within a time period which provides both in the same location, e.g., the stomach, at the same time).
  • the delivery agent passes though the mucosal barriers of the gastrointestinal tract and is absorbed into the blood stream where it can be detected in the plasma and/or blood of subjects.
  • the level of delivery agent in the bloodstream as measured in the plasma and/or blood is dose-dependent.
  • One preferred pharmaceutical composition of the inventions comprises a combination of insulin and a delivery agent in a suitable pharmaceutical carrier or excipient as understood by practitioners in the art. It is preferred that the dosage form of the pharmaceutical composition of the present invention is a solid dosage form, such as a gelatin capsule or a tablet.
  • the ratio of portal (unmodified) insulin concentration to systemic (unmodified) insulin concentration approaches in human diabetic patients approaches that which is obtained in normal healthy humans.
  • the chronic administration of oral dosage forms of the present invention result in a higher portal insulin concentration and lower systemic insulin concentration over time than that obtained with an equi-effective dose of insulin administered subcutaneously (i.e., which provide similar control of blood glucose levels).
  • Transient peaks in insulin levels that may occur by virtue of the oral administration of insulin in accordance with the present invention is not believed to be associated with vascular diseases.
  • insulin refers to insulin from a variety of sources. Naturally occurring insulin and structurally similar bioactive equivalents (insulin analogues including short acting and analogues with protracted action) can be used. Insulin useful in the invention can be may be obtained by isolating it from natural source, such as different species of mammal. For example, animal insulin preparations extracted from bovine or porcine pancreas can be used. Insulin analogues, fragments, mimetics or polyethylene glycol (PEG)-modified derivatives of these compounds, derivatives and bioequivalents thereof can also be used with the invention.
  • PEG polyethylene glycol
  • the insulin used in the present invention may be obtained by chemically synthesizing it using protein chemistry techniques such as peptide synthesis, or by using the techniques of molecular biology to produce recombinant insulin in bacteria or eukaryotic cells.
  • the physical form of insulin may include crystalline and/or amorphous solid forms.
  • dissolved insulin may be used.
  • Other suitable forms of insulin, including, but not limited to, synthetic forms of insulin, are described in U.S. Patents Nos. 4,421,685, 5,474,978, and 5,534,488, the disclosure of each of which is hereby incorporated by reference in its entirety.
  • the most preferred insulin useful in the pharmaceutical compositions and methods of the present invention is human recombinant insulin optionally having counter ions including zinc, sodium, calcium and ammonium or any combination thereof.
  • Human recombinant insulin can be prepared using genetic engineering techniques that are well known in the art. Recombinant insulin can be produced in bacteria or eukaryotic cells. Functional equivalents of human recombinant insulin are also useful in the invention. Recombinant human insulin can be obtained from a variety of commercial sources. For example, insulin (Zinc, human recombinant) can be purchased from Calbiochem (San Diego, CA).
  • human recombinant Zinc-Insulin Crystals Proinsulin Derived (Recombinant DNA Origin) USP Quality can be obtained from Eli Lilly and Company (Indianapolis, IN). All such forms of insulin, including insulin analogues (including but not limited to Insulin Lispro, Insulin Aspart, Insulin Glargine, and Insulin Detemir) are deemed for the purposes of this specification and the appended claims are considered to be encompassed by the term "insulin.”
  • the present invention also provides compositions of recombinant human zinc insulin and a delivery agent as a drug for oral administration of insulin in humans.
  • the insulin is a modified insulin, such as that conjugated with an oligomer such as that described in U.S. Patent No. 6,309,633 and/or which not has been subjected to amphiphilic modification such as that described in U.S. Patent Nos. 5,359,030; 5,438,040; and/or 5,681,811.
  • the conjugated (modified) insulin may be incorporated into the oral formulations of the present invention in addition to or in the absence of any of the types of insulin described above, as well as with other insulin analogues.
  • the oral formulations include the modified insulin either with or without a pharmaceutically acceptable delivery agent that facilitates absorption of said insulin from the gastrointestinal tract.
  • the oral dosage form comprise a therapeutically effective amount of unmodified insulin, i.e., a pharmacologically or biologically effective amount, or an amount effective to accomplish the purpose of insulin.
  • the dose of insulin administered should preferably be in such an amount that, upon oral administration, it results in a measurable and statistically significant reduction in blood glucose levels in normal healthy human subjects.
  • the amount can be less than a pharmacologically or biologically effective amount when the composition is used in a dosage unit form, such as a tablet, because the dosage unit form may contain a multiplicity of delivery agent/biologically or chemically active agent compositions or may contain a divided pharmacologically or biologically effective amount.
  • the total effective amounts can then be administered in cumulative units containing, in total, pha ⁇ nacologically, biologically or chemically active amounts of biologically or pharmacologically active agent.
  • the total amount of insulin to be used can be determined by those skilled in the art.
  • the amount of insulin is an amount effective to accomplish the purpose of the particular active agent.
  • the amount in the composition is a therapeutically effective dose, i.e., a pharmacologically or biologically effective amount.
  • the amount can be less than a pharmacologically or biologically effective amount when the composition is used in a dosage unit form, such as a capsule, a tablet or a liquid, because the dosage unit form may contain a multiplicity of delivery agent/biologically or chemically active agent compositions or may contain a divided pharmacologically or biologically effective amount.
  • the total effective amounts can then be administered in cumulative units containing, in total, pharmacologically or biologically or chemically active amounts of biologically or pharmacologically active agent.
  • Preferred insulin doses when dosed in combination with the delivery agents described herein, are about 50 to about 600 insulin Units USP (from about 2 to about 23mg), preferably from about 100 Units (3.8 mg) to about 450 Units (15.3 mg), more preferably from about 200 Units (7.66 mg) to about 350 Units (13.4 mg), and still more preferably about 300 Units (11.5 mg), based on the accepted conversion of factor of 26.11 Units per mg.
  • the oral dosage forms of the invention facilitate the oral delivery of insulin, and after insulin is absorbed into the bloodstream, the composition produces a maximal decrease in blood glucose in treated patients from about 20 to about 60 minutes after oral administration.
  • the pharmaceutical composition produces a maximal decrease in blood glucose in treated patients from about 30 to about 50 minutes post oral administration. More particularly, the pharmaceutical composition produces a maximal decrease in blood glucose in treated patients at about 40 minutes after oral administration.
  • human diabetic patients show a maximal decrease in blood glucose by at least 10% within one hour post oral administration. In another embodiment, human diabetic patients show a maximal decrease in blood glucose by at least 20% within one hour post oral administration, alternatively, at least 30% within one hour post oral administration.
  • One goal of the present invention is to provide oral compositions of insulin that facilitate achieving close to normal levels of blood glucose throughout the 24-hour daily cycle.
  • the pharmaceutical composition includes insulin or an insulin analog as the active agent and a delivery agent in an amount effective to achieve a fasting blood glucose concentration from about 90 to about 110 mg/dl.
  • the pharmaceutical composition includes insulin or an insulin analog as the active agent and a delivery agent in an amount effective to achieve a fasting blood glucose concentration from about 95 to about 105 mg/dl, more preferably, the subject manifests fasting blood glucose concentrations at about 100 mg/dl.
  • the present invention provides oral compositions of insulin that prevent or control very high levels of blood glucose from being reached and/or sustained. More particularly, the present invention provides compositions which facilitate achieving normal levels of blood glucose after a meal has been consumed, i.e., post-prandial.
  • the pharmaceutical composition includes insulin as the active agent and a delivery agent in an amount effective to achieve a post-prandial blood glucose concentration from about 130 to about 170 mg/dl.
  • the pharmaceutical composition includes insulin or an insulin analog as the active agent and a delivery agent in an amount effective to achieve a post- prandial blood glucose concentration from about 140 to about 160 mg/dl, more preferably, the subject manifests fasting blood glucose concentrations at less than about 160 mg/dl.
  • the present invention provides pharmaceutical compositions for oral administration which includes insulin or an insulin analog as the active agent and a delivery agent in an amount effective to achieve pre-prandial (before a meal is consumed) blood glucose concentration from about 95 to about 125 mg/dl.
  • the present invention provides pharmaceutical compositions for oral administration which includes insulin or an insulin analog as the active agent and a delivery agent in an amount effective to achieve pre-prandial blood glucose concentration from about 100 to about 120 mg/dl.
  • the present invention provides pharmaceutical compositions for oral administration which include insulin as the active agent and a delivery agent in an amount effective to achieve blood glucose concentrations within the normal range during the evening period from about 70 to about 120 mg/dl.
  • the present invention provides pharmaceutical compositions for oral administration which include insulin or an insulin analog as the active agent and a delivery agent in an amount effective to achieve blood glucose concentrations at 3 AM from about 80 to about 120 mg/dl.
  • the methods and pharmaceutical compositions provide the pharmacokinetic parameters set forth in United States Provisional Applications Nos. 60/346,746 and 60/347,312, the disclosure of each of which is incorporated herein by reference.
  • the amount of delivery agent necessary to adequately deliver insulin into the blood stream of a subject needing the therapeutic effect of insulin can vary depending on one or more of the following; chemical structure of the particular delivery agent; the nature and extent of interaction of insulin and the delivery agent; the nature of the unit dose, i.e., solid, liquid, tablet, capsule, suspension; the concentration of delivery agent in the GI tract, the feeding state of the subject, the diet of the subject, the heath of the subject and the ratio of delivery agent to insulin.
  • the oral dosage forms of the present invention comprise a mixture of insulin and a delivery agent, e.g., monosodium N-(4-chlorosalicyloyl)-4-aminobutyrate (4-CNAB), a novel compound discovered by Emisphere Technologies, Inc., or separately containing insulin and the delivery agent.
  • a delivery agent e.g., monosodium N-(4-chlorosalicyloyl)-4-aminobutyrate (4-CNAB), a novel compound discovered by Emisphere Technologies, Inc.
  • the oral dosage forms described herein are orally administered as described herein in combination with an additional therapy to treat diabetes, impaired glucose tolerance, or to achieve glucose homeostasis, said additional therapy comprising, for example, an additional drug such as sulfonylurea, a biguanide, an alpha- glucosidase, insulin delivered via a different pathway (e.g., parenteral insulin), and/or an insulin sensitizer.
  • an additional drug such as sulfonylurea, a biguanide, an alpha- glucosidase, insulin delivered via a different pathway (e.g., parenteral insulin), and/or an insulin sensitizer.
  • the oral dosage forms described herein reduce the likelihood of hypoglycemic events, mainly because of two reasons.
  • the short peak of insulin e.g., as shown in the appended examples shows that, even if insulin were to reach high peripheral levels, the peak drops precipitously.
  • the pharmaceutical composition comprises insulin as the active agent and the compound 4-CNAB as a delivery agent to facilitate the oral delivery of insulin, and, after insulin is absorbed into the bloodstream, the composition produces a maximal decrease in C-peptide concentration in treated patients from about 80 and about 120 minutes post oral administration. More particularly, the composition produces a maximal decrease in C-peptide concentration in treated patients from about 90 and about 110 minutes post oral administration.
  • Absorption of insulin can be detected in subjects treated with the pharmaceutical compositions of the present invention by monitoring the plasma levels of insulin after treatment.
  • the time it takes for an active agent to reach a peak in the bloodstream may depend on many factors such as the following: the nature of the unit dose, i.e., solid, liquid, tablet, capsule, suspension; the concentration of active agent and delivery agent in the GI tract; the feeding state of the subject; the diet of the subject; the health of the subject and the ratio of active agent to the delivery agent.
  • the pharmaceutical composition includes the compound 4-CNAB as the delivery agent and insulin as the active agent
  • the composition provides a peak plasma insulin concentration from about 0.1 to about 1 hour after oral administration.
  • the composition provides a peak plasma insulin concentration from about 0.2 to about 0.6 hours after oral administration. In a preferred embodiment, the composition provides a peak plasma insulin concentration from about 0.3 to about 0.4 hours after oral administration. In another embodiment, the composition provides a peak plasma insulin concentration within about 1 hour after oral administration.
  • the pharmaceutical composition comprises insulin as the active agent and the compound 4-CNAB as a delivery agent to facilitate the oral delivery of insulin, and after insulin is absorbed into the bloodstream, the plasma insulin levels in treated patients peak at about 20 minutes post oral administration with a second peak at about 105 minutes.
  • the delivery agents used in the invention have the following structure:
  • X is one or more of hydrogen, halogen, hydroxyl or C ⁇ -C 3 alkoxy, and R is substituted or unsubstituted C ⁇ -C 3 alkylene, substituted or unsubstituted C ⁇ -C 3 alkenylene.
  • the delivery agents of the invention preferably have the following structure:
  • X is halogen
  • R is substituted or unsubstituted C ⁇ -C 3 alkylene, substituted or unsubstituted C ⁇ -C 3 alkenylene.
  • the pharmaceutical composition includes a delivery agent wherein X is chlorine and R is C 3 alkylene.
  • the pharmaceutical composition includes the compound 4- [(4-chloro, 2-hydrqxybenzoyl)amino]butanoic acid as a delivery agent for the oral delivery of insulin, preferably the monosodium salt thereof.
  • the delivery agents may be in the form of the carboxylic acid or salts thereof.
  • Suitable salts include, but are not limited to, organic and inorganic salts, for example alkali-metal salts, such as sodium, potassium and lithium; alkaline-earth metal salts, such as magnesium, calcium or barium; ammonium salts; basic amino acids, such as lysine or arginine; and organic amines, such as dimethylamine or pyridine.
  • the salts are sodium salts.
  • the salts may be mono- or multi-valent salts, such as monosodium salts and di-sodium salts.
  • the salts may also be solvates, including ethanol solvates, and hydrates.
  • suitable delivery agents that can be used in the present invention include those delivery agents described United States Patents Nos. 5,650,386, 5,773,647, 5,776,888, 5,804,688, 5,866,536, 5,876,710, 5,879,681, 5,939,381, 5,955,503, 5,965,121,5,989,539, 5,990,166, 6,001,347, 6,051,561, 6,060,513, 6,090,958, 6,100,298, 5,766,633, 5,643,957, 5,863,944, 6,071,510 and 6,358,504, the disclosure of each of which is incorporated herein by reference. Additional suitable delivery agents are also described in International Publications Nos.
  • Salts of the delivery agent compounds of the present invention may be prepared by methods known in the art.
  • sodium salts may be prepared by dissolving the delivery agent compound in ethanol and adding aqueous sodium hydroxide.
  • the compounds described herein may be derived from amino acids and can be readily prepared from amino acids by methods known by those with skill in the art based upon the present disclosure and the methods described in International Publications Nos. WO 96/30036, WO 97/36480, WO 98/34632 and WO 00/07979, and in United States Patents Nos. 5,643,957 and 5,650,386, the disclosure of each of which is incorporated herein by reference.
  • the compounds may be prepared by reacting the single amino acid with the appropriate acylating or amine-modifying agent, which reacts with a free amino moiety present in the amino acid to form amides.
  • Protecting groups may be used to avoid unwanted side reactions as would be known to those skilled in the art.
  • the delivery agents may also be prepared by the methods of International Publications Nos. WO 02/02509 and WO 03/057650), the disclosures of which is incorporated herein by reference.
  • the delivery agents may also be prepared by alkylation of the appropriate salicylamide according to the methods of International Publication No. WO 00/46182, the disclosure of which is incorporated herein by reference.
  • the salicylamide may be prepared from salicylic acid via the ester by reaction with sulfuric acid and ammonia.
  • poly amino acids and peptides comprising one or more of these compounds may be used.
  • An amino acid is any carboxylic acid having at least one free amine group and includes naturally occurring and synthetic amino acids.
  • Poly amino acids are either peptides (which are two or more amino acids joined by a peptide bond) or are two or more amino acids linked by a bond formed by other groups which can be linked by, e.g., an ester or an anhydride linkage.
  • Peptides can vary in length from dipeptides with two amino acids to polypeptides with several hundred amino acids.
  • the delivery agent compound may be purified by recrystallization or by fractionation on one or more solid chromatographic supports, alone or linked in tandem.
  • Suitable recrystallization solvent systems include, but are not limited to, ethanol, water, heptane, ethyl acetate, acetonitrile, methanol and tetrahydrofuran and mixtures thereof. Fractionation may be performed on a suitable chromatographic support such as alumina, using methanol/n-propanol mixtures as the mobile phase; reverse phase chromatography using trifluoroacetic acid/ acetonitrile mixtures as the mobile phase; and ion exchange chromatography using water or an appropriate buffer as the mobile phase.
  • anion exchange chromatography preferably a 0-500 mM sodium chloride gradient is employed.
  • the delivery agent passes though the mucosal barriers of the GI tract and is absorbed into the blood stream where it can be detected in the plasma of subjects.
  • the level of delivery agent in the bloodstream as measured in the plasma is dose-dependent.
  • the delivery agent facilitates the absorption of the drug (active agent) administered therewith (either in the same dosage form, or simultaneously therewith), or sequentially (in either order, as long as both the delivery agent and the drug are administered within a time period which provides both in the same location, e.g., the stomach, at the same time).
  • a peak plasma concentration (C max ) of the delivery agent achieved after oral administration is preferably from about 10 to about 250,000 ng/ml, after oral administration, preferably from about 100 to about 125,000, and preferably the peak plasma concentration of the delivery agent is from about 1,000 to about 50,000 ng/ml, after oral administration. More preferably, the peak plasma concentration of the delivery agents of the present invention is from about 5,000 to about 15,000 ng/ml, after oral administration.
  • the time it takes for the delivery agent to reach a peak in the bloodstream may depend on many factors such as the following: the nature of the unit dose, i.e., solid, liquid, tablet, capsule, suspension; the concentration of delivery agent in the GI tract; the feeding state of the subject; the diet of the subject; the health of the subject and the ratio of delivery agent to the active agent.
  • the delivery agents of the present invention are rapidly absorbed from the gastrointestinal tract when orally administered in an immediate release dosage form, and preferably provide a peak plasma concentration within about 0.1 to about 8 hours after oral administration, and preferably at about 0.1 to about 3 hours after oral administration.
  • the t max of the delivery agent occurs at about 0.3 to about 1.5 hours after oral administration. In certain embodiments, the delivery agent achieves a t max within about 2 hours after oral administration, and most preferably, within about 1 hour after oral administration.
  • the amount of delivery agent in the present composition is a delivery effective amount and can be determined by methods known to those skilled in the art.
  • the amount of delivery agent necessary to adequately deliver an active agent into the blood stream of a subject needing the therapeutic effect of that active agent may vary depending on one or more of the following; the chemical nature of the active agent; the chemical structure of the particular delivery agent; the nature and extent of interaction from about the active agent and delivery agent; the nature of the unit dose, i.e., solid, liquid, tablet, capsule, suspension; the concentration of delivery agent in the GI tract; the feeding state of the subject; the diet of the subject; the health of the subject and the ratio of delivery agent to the active agent.
  • the amount of the delivery agent preferred for the pharmaceutical composition is from about 1 mg to about 2,000 mg delivery agent, more preferably from about 1 mg to about 800 mg of said delivery agent, more preferably from about 50 mg to about 700 mg of said delivery agent, even more preferably from about 70 mg to about 700 mg of said delivery agent, still more preferably from about 100 to about 600 mg.
  • the delivery agent is 4-CNAB. Since the amount of delivery agent required to deliver a particular active agent is variable and the amount of active agent required to produce a desired therapeutic effect is also a variable, the ratio of active agent to delivery agent may vary for different active agent/delivery agent combinations. In certain preferred embodiments of the invention where the oral pharmaceutical composition includes insulin as the active agent and the delivery agent is the compound 4-CNAB, the amount of the delivery agent included in the pharmaceutical composition may be from about 100 mg to about 600 mg of said delivery agent.
  • the pharmaceutical composition includes insulin as the active agent and the delivery agent is the monosodium salt of 4-CNAB, the ratio of insulin [Units] to delivery agent [mg] ranges from 10:1 [Units/mg] to 1:10 [Units/mg], preferably, the ratio of insulin [Units] to delivery agent [mg] ranges from 5:1 [Units/mg] to 0.5:1 [Units/mg].
  • Preferred insulin doses in a single administration are about 5 to about 1000 insulin units USP, preferably from about 50 to about 400, more preferably from about 150 to about 400, and still more preferably from about 150 to about 300 units.
  • the optimum ratio of insulin to delivery agent can vary depending on the delivery agent. Optimizing the ratio of insulin to delivery agent is within the knowledge of one skilled in the art.
  • the composition provides a peak plasma delivery agent concentration within about 0.1 to about 3 hours after oral administration.
  • the peak plasma concentration of delivery agent attained is from about 8,000 to about 37,000 ng/ml.
  • Emisphere Technologies, Inc. disclosed structures of various delivery agents, comparisons of their effectiveness of absorption and effectiveness of delivery, the preparation of the preferred delivery agent 4-CNAB, its preparation for human studies, and data regarding previous non-clinical and clinical studies involving the delivery agent 4-CNAB.
  • the delivery agent may be used directly by mixing with the unmodified insulin prior to administration, either in dry powder form or wet granulated together. To this mixture, other pharmaceutically acceptable excipients may be added. The mixture may be then tableted or placed into gelatin capsules containing a unit dose of the active agent and the delivery agent. Alternatively, the delivery agent/insulin mixture may be prepared as an oral solution or suspension. The delivery agent and insulin do not need to be mixed together prior to administration, such that, in certain embodiments, the unit dose of insulin (with or without other pha ⁇ naceutically acceptable excipients) is orally administered without the delivery agents of this invention, and the delivery agent is separately orally administered (with or without other pharmaceutically acceptable excipients) before, after, or simultaneously with the insulin.
  • the oral dosage forms of the present invention are solid.
  • the unmodified insulin in dry powder form is stable, and in certain prefe ⁇ ed embodiments is simply mixed in a desirable ratio with the delivery agent.
  • the dry powder mixture may then be filled into gelatin capsules, with or without optional pharmaceutical excipients.
  • the unmodified insulin in dry powder form may be mixed with the delivery agent together with optional pharmaceutical excipients, and the mixture may be tableted in accordance with standard tableting procedures known to those having ordinary skill in the art.
  • the dosage forms of the present invention may be produced by first dissolving insulin and the delivery agent into one solution or separate solutions.
  • the solvent will preferably be an aqueous solution, but organic solvents or aqueous organic solvent mixtures may be used when necessary to solubilize the delivery agent. If two solutions are used, the proportions of each necessary to provide the correct amount of either insulin or delivery agent are combined and the resulting solution may be dried, by lyophilization or equivalent means.
  • the oral dosage form may be dried and rehydrated prior to oral administration.
  • the administration mixtures may be prepared, e.g., by mixing an aqueous solution of the delivery agent with an aqueous solution of insulin just prior to administration. Alternatively, the delivery agent and insulin can be admixed during the manufacturing process.
  • the solutions may optionally contain additives such as phosphate buffer salts, citric acid, acetic acid, gelatin, and gum acacia.
  • Stabilizing additives may be incorporated into the delivery agent solution. With some drugs, the presence of such additives promotes the stability and dispersibility of the agent in solution.
  • the stabilizing additives may be employed at a concentration ranging from about 0.1 and 5% (W/N), preferably about 0.5% (WN).
  • stabilizing additives include gum acacia, gelatin, methyl cellulose, polyethylene glycol, carboxylic acids and salts thereof, and polylysine.
  • the prefe ⁇ ed stabilizing additives are gum acacia, gelatin and methyl cellulose.
  • the amount of delivery agent in the present composition is a delivery effective amount and can be determined for any particular delivery agent/active agent combination by methods known to those skilled in the art.
  • the oral dosage forms of the present invention containing a mixture of insulin and the delivery agent, e.g., 4-C ⁇ AB, or separately containing insulin and the delivery agent, may include additional materials known to those skilled in the art as pharmaceutical excipients. Any excipient or ingredient, including pharmaceutical ingredients or excipients.
  • Such pharmaceutical excipients include, for example, the following: Acidifying agents (acetic acid, glacial acetic acid, citric acid, fumaric acid, hydrochloric acid, diluted hydrochloric acid, malic acid, nitric acid, phosphoric acid, diluted phosphoric acid, sulfuric acid, tartaric acid); Aerosol propellants (butane, dichlorodifluoro- methane, dichlorotetrafluoroethane, isobutane, propane, trichloromonofluoromethane); Air displacements (carbon dioxide, nitrogen); Alcohol denaturants (denatonium benzoate, methyl isobutyl ketone, sucrose octacetate); Alkalizing agents (strong ammonia solution, ammonium carbonate, diethanolamine, diisopropanolamine, potassium hydroxide, sodium bicarbonate, sodium borate, sodium carbonate, sodium hydroxide, trolamine); Anticaking agents (see glidant); Antifo
  • the insulin present in the dosage unit form is absorbed into the circulation.
  • the bioavailability of the insulin is readily assessed by measuring a known pharmacological activity in blood, e.g., decreased blood glucose. Further physiologic effects of the insulin can be measured using tests, for example, measurement of plasma C-peptide concentration. Alternately, the circulating levels of the insulin itself can be measured directly. Similarly, levels of 4-CNAB or other delivery agent in the blood can be measured.
  • the reaction was poured into a separatory funnel and washed with 2N HC1 and an emulsion formed.
  • the emulsion was left standing for two days and was then filtered through celite in a fritted glass funnel. The filtrate was put back in a separatory funnel to separate the layers.
  • the organic layer was dried over sodium sulfate, which was then filtered off and the filtrate concentrated by rotary evaporation.
  • the resulting solid material was hydrolyzed with 2N NaOH, stored overnight under refrigeration, and then hydrolyzing resumed.
  • the solution was acidified with 2N HC1 and the solids that formed were isolated, dried under vacuum, and recrystallized twice using methanol/water. Solids precipitated out overnight and were isolated and dried.
  • 4-CNAB for the human dosings (Monosodium N-(4-chlorosalicyloyl)-4-amino-butyrate) was made under good manufacturing practices (GMP) conditions by Regis Technologies, Inc. (Morton Grove, IL) according to the methods of International Publication No. WO 00/46182 except that the starting material 4-chlorosalicylic acid (purchased from Ihara Chemical Industry Co. Inc, Ltd., Tokyo, Japan and Aapin Chemicals Ltd., Oxfordshire, UK) was used and converted to the amide via a methyl ester using 0.14 equivalents sulfuric acid in methanol and then about 4 equivalents ammonia in methanol.
  • the alkylating agent used was ethyl-4-bromobutyrate.
  • the monosodium salt of 4-CNAB was made according to the following method on a 40 kilogram scale.
  • the flask was equipped with an overhead sti ⁇ er, a thermocouple temperature read out, a reflux condenser and a heating mantle, and was placed under nitrogen.
  • Reagent grade acetone 13 L was added to the reactor and the mixture was agitated.
  • the 4-CNAB/acetone mixture was heated to 50° C to dissolve any solids. A hazy brown solution was achieved.
  • the 50° C solution was pumped through a warm pressure filter (dressed with Whatman #1 filter paper, ⁇ 5 microns, 18.5 sq. in. area) into a clean 22 L reactor to remove sodium chloride and other insolubles.
  • the pressure dropped across the filter to about 20 psig at the end of filtration.
  • the reactor containing the clear yellow filtrate was agitated and heated. At 50° C the reactor was removed from heat.
  • the insulin for the subcutaneous injection was HUMULIN " R injection insulin from Eli Lilly and Company (Indianapolis, IN).
  • All capsules containing 200 mg 4-CNAB and 150 insulin units USP were prepared as follows. First, the total amount of delivery agent material necessary for filling the delivery agent alone capsules and the delivery agent plus insulin composition capsules was prepared by weighing 3160 g of 4-CNAB. The 3160 g 4-CNAB was then milled in a Quadro comil, model iP7Smill with screen number 2A 050 G 037 19 136 (1270 micron). Next, 1029 g of the milled 4-CNAB was passed through a #35 mesh screen. Then, the pass through screened material was transfe ⁇ ed into a 4 quart shell and blended using for example, a V blender, at 25 rpm for 10.2 minutes.
  • the resultant blended material was used to fill capsules.
  • a Fast Cap Capsule Filler was used with a size 3 Fast Cap Encapsulation tray.
  • the empty capsules weighed approximately 48 mg each and were filled with an average fill weight of 205.6 mg of 4-CNAB alone. Thus, the dose of the delivery agent alone capsules was 205.6 mg.
  • the insulin compositions were prepared by first dispensing 31.8 g of recombinant human zinc crystalline insulin (Potency 26.18 Units per mg) (proinsulin derived (recombinant DNA origin) USP quality) from Eli Lilly and Company (Indianapolis, IN) into an appropriately sized plastic bag. Next, sequential 30 g additions of the milled and screened 4-CNAB were added to the bag until approximately 510 g had been added. The bag was thoroughly mixed after each 30 g addition of 4-CNAB by shaking and inversion.
  • the 541.8 g mixture of insulin and 4-CNAB was transfe ⁇ ed to a V blender and mixed again at 25 rpm for 10.2 minutes.
  • the remaining 4-CNAB was added to the blender and the entire mixture was mixed in the blender at 25 rpm for 10.2 minutes.
  • the resulting composition was dispensed as described above into empty capsules.
  • the final capsules contained an average of 5.1 mg insulin (equivalent to 150 units insulin) and 200.5 mg of 4-CNAB or a ratio of 1:57.3, insulin: 4-CNAB. Multiple samples of the final blend were run on HPLC to verify uniformity and were found to be uniform.
  • the present invention comprising compositions of insulin and the delivery agent 4-CNAB was evaluated for safety and toxicity in a non-clinical program that included pha ⁇ nacological screening, pharmacokinetic profiling, and toxicity assessments in rats and monkeys.
  • animal physiological responses to 4-CNAB alone and to Insulin/4-CNAB were comparable.
  • Pharmacokinetic studies in mice, rats and monkeys have shown that 4-CNAB is absorbed rapidly following oral administration, and subsequently cleared from the body. 4-CNAB did not demonstrate potential activity in any of the primary molecular targets evaluated in receptor binding screening assays.
  • Four genotoxicity studies have been conducted with 4-CNAB, with no positive findings. Based on 14-day oral repeated dose toxicity studies, the NOAEL (No-Adverse Effect Level) was estimated to be 500 mg/kg in Sprague-Dawley rats, and 400 mg/kg in rhesus monkeys.
  • the starting insulin dose of 150 insulin Units USP (which is about 7-fold lower than the 15 U/kg no effect dose in monkey) was selected.
  • This example describes the procedure for preparing Insulin/4-CNAB capsules.
  • the 4-CNAB as prepared above was first screened through a 35 mesh screen. The required amount of the screened 4-CNAB was weighed and was kept in a covered weighing boat. The required amount of insulin was weighed and was kept in a covered weighing boat.
  • the insulin from above was screened through the 35 mesh screen onto the same mortar, and approximately 2.0 grams of the 4-CNAB from above was screened on top of the insulin using the same 35 mesh screen.
  • the contents of the mortar was mixed by light trituration for about 3 minutes with a glass mortar, with a spatula used for scraping, as necessary.
  • the 4-CNAB from above was continued to be screened through the same 35 mesh screen in small portions equivalent to the volume of material in the mortar. After each addition, the contents of the mortar were mixed for about 3 minutes.
  • the contents of the mortar was mixed by light trituration with a glass mortar for about 2 minutes, with a spatula used for scraping, as necessary.
  • the final blend was transfe ⁇ ed into a weighing boat for capsule filling and hand-filled into the capsules.
  • the oral insulin capsule(s) described herein were orally administered to twenty human subjects with diabetes at night before going to sleep.
  • this example reports the results of an open-label, single- dose, crossover study comparing the safety of orally administered Insulin/4-CNAB formulation in two groups of subjects with type 2 diabetes mellitus — one in the fasting state and one with a standard meal.
  • the objectives were (1) to compare the safety, pharmacokinetics and pharmacodynamics of orally administered Insulin/4-CNAB in fasting type 2 diabetic subjects, and (2) to compare blood glucose, insulin and C-peptide levels after a standard meal with diet or regular medication with blood glucose, insulin and C-peptide levels after a standard meal with Insulin/4-CNAB.
  • the focus of this example is the assessment of the safety, pharmacokinetics and phannacodynamics of insulin/4-CNAB, administered orally at bedtime, to type 2 diabetic subjects.
  • the purpose of the study was to determine if the administration of oral insulin at bedtime could exert effects on overnight-fasting glucose homeostasis and insulin secretion.
  • the postulated mode of action e.g., suppressing the liver production of glucose, and thus preventing beta cell damage or even death, leading to exacerbation of dysfunction of insulin production was the basis for the design of the study.
  • Group 1 twelve (12) type 2 diabetic subjects: (a) oral insulin/4-CNAB - fasted subjects, and (b) empty capsule — fasted subjects.
  • Group 2 twelve (12) type 2 diabetic subjects: (a) standard meal with regular medication, and (b) oral human insulin 4-CNAB prior to standard meal.
  • the subjects ate their regular dinner at home, as every evening, between the hours of 7:00 and 8:00 P.M. If the subjects usually took medication for the diabetes (e.g., Metformin or Acarbose) in the evening, they took their usual doses. At 11 :00 p.m. (at least two hours after dinner), the subjects took one oral insulin dose that contained the following ingredients: 300 mg 4-CNAB and insulin according to the dose (200-400 U) that the subject received during the first phase of the trial. If the subject had received 200 U insulin in the first phase of the trial and there was no drop in blood glucose level ( ⁇ 15% reduction), that subject now received 300 U of insulin.
  • the diabetes e.g., Metformin or Acarbose
  • the subjects' blood glucose levels were checked with a glucometer before the subjects took the medication. In addition, blood was taken for further blood glucose levels, insulin and C- peptide. Orange juice was readily available for treatment in the unlikely event of hypoglycemia.
  • the subjects During sleep, the subjects wore a Glucowatch (which monitors blood glucose and measures and records blood glucose levels at regular intervals), which is equipped with an alarm that is triggered when blood glucose levels reach predetermined blood glucose levels (hypoglycemic levels) determined by the investigator or patient.
  • the subjects' blood glucose levels were checked with the glucometer, and additional blood samples were taken for further blood glucose levels, insulin and C-peptide.
  • the blood samples from the night before were stored in the refrigerator at home, and in the morning the nurse brought the samples of blood (from the night and the morning) to the lab for analysis.
  • Figures 1-5 The results of the nighttime oral insulin study reported as the example herein (fasting blood glucose, insulin and C-peptide measured at approximately 7:00 a.m. and compared to the patient's own baseline levels) are set forth in Figures 1-5. The results are reported by patient with numeric values in Figures 1 and 2 ( ⁇ U/ml), and are graphically represented in Figures 3-5.
  • Figure 1 shows data for blood glucose, insulin and C-peptide collected in the morning after nighttime dosing of insulin/4-CNAB for each subject compared to that subject's own baseline levels
  • Figure 2 shows data for insulin and C-peptide collected at night prior to nighttime dosing of insulin and 4- CNAB for each subject.
  • Figures 3-5 are bar graphs showing the effect of nighttime dosing of insulin/4-CNAB on blood glucose concentration, blood C-peptide concentration and blood insulin concentration, respectively.
  • bedtime insulin caused a reduction in plasma insulin level the morning after ingestion of the insulin/4-CNAB capsule compared with that measured the two mornings when the subjects came to the clinic for the first stage of the trial.
  • the bedtime dosing of oral insulin resulted in suppressed overnight fasting insulin demand and may improve insulin sensitivity (unchanged fasting glycemia with reduced systemic hyperinsulinemia).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Diabetes (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Endocrinology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Emergency Medicine (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicinal Preparation (AREA)

Abstract

L'invention concerne un procédé de protection d'un mammifère souffrant d'une diminution de la tolérance au glucose ou du diabète sucré précoce contre le développement du diabète insulino-dépendant ou explicite consistant à administrer une dose oralement efficace d'une formulation pharmaceutique renfermant une insuline le soir, p.ex. au moment où le patient va se coucher ou peu avant.
PCT/US2004/000273 2003-01-06 2004-01-06 Therapie d'administration orale d'insuline le soir WO2004062587A2 (fr)

Priority Applications (8)

Application Number Priority Date Filing Date Title
NZ541058A NZ541058A (en) 2003-01-06 2004-01-06 Night-time oral insulin therapy
CA2511530A CA2511530C (fr) 2003-01-06 2004-01-06 Therapie d'administration orale d'insuline le soir
US10/541,433 US7384914B2 (en) 2003-01-06 2004-01-06 Night-time oral insulin therapy
JP2006500813A JP5452843B2 (ja) 2003-01-06 2004-01-06 夜間の経口インスリン治療
ES04700388.4T ES2465496T3 (es) 2003-01-06 2004-01-06 Terapia de insulina oral nocturna
AU2004204727A AU2004204727B8 (en) 2003-01-06 2004-01-06 Night-time oral insulin therapy
EP04700388.4A EP1592438B1 (fr) 2003-01-06 2004-01-06 Therapie d'administration orale d'insuline le soir
HK06105431.0A HK1085133A1 (en) 2003-01-06 2006-05-09 Night-time oral insulin therapy

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US43819503P 2003-01-06 2003-01-06
US43845103P 2003-01-06 2003-01-06
US60/438,451 2003-01-06
US60/438,195 2003-01-06
US47896703P 2003-06-16 2003-06-16
US60/478,967 2003-06-16

Publications (2)

Publication Number Publication Date
WO2004062587A2 true WO2004062587A2 (fr) 2004-07-29
WO2004062587A3 WO2004062587A3 (fr) 2005-04-28

Family

ID=32719186

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/000273 WO2004062587A2 (fr) 2003-01-06 2004-01-06 Therapie d'administration orale d'insuline le soir

Country Status (9)

Country Link
US (1) US7384914B2 (fr)
EP (1) EP1592438B1 (fr)
JP (1) JP5452843B2 (fr)
AU (1) AU2004204727B8 (fr)
CA (1) CA2511530C (fr)
ES (1) ES2465496T3 (fr)
HK (1) HK1085133A1 (fr)
NZ (1) NZ541058A (fr)
WO (1) WO2004062587A2 (fr)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005112937A1 (fr) 2004-05-19 2005-12-01 Emisphere Technologies, Inc. Formulations d’acyclovir
WO2006072070A2 (fr) 2004-12-29 2006-07-06 Emisphere Technologies, Inc. Formulations pharmaceutiques contenant des sels de gallium
JP2008509145A (ja) * 2004-08-03 2008-03-27 エミスフィアー テクノロジーズ インコーポレイテッド 抗糖尿病性経口インスリン−ビグアニドの組み合わせ
EP2279732A2 (fr) 2004-05-14 2011-02-02 Emisphere Technologies, Inc. Composés et compositions pour la distribution d'agents actifs
WO2011017346A2 (fr) 2009-08-03 2011-02-10 Emisphere Technologies, Inc. Composition de naproxène à action rapide avec effets gastro-intestinaux réduits
US7939494B2 (en) 2002-02-20 2011-05-10 Emisphere Technologies, Inc. Method for administering GLP-1 molecules
US8110547B2 (en) 2005-01-12 2012-02-07 Emisphere Technologies, Inc. Compositions for buccal delivery of parathyroid hormone
US8324156B2 (en) 2003-08-22 2012-12-04 Emisphere Technologies, Inc. Oral insulin therapies and protocol
US8771712B2 (en) 2006-05-09 2014-07-08 Emisphere Technologies, Inc. Topical administration of acyclovir
US8785381B2 (en) 2003-12-19 2014-07-22 Emisphere Technologies, Inc. Oral GLP-1 formulations

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1605895A4 (fr) * 2003-03-06 2011-08-24 Emisphere Tech Inc Insulinotherapies orales et protocole
JP2008542235A (ja) * 2005-05-25 2008-11-27 ノボ・ノルデイスク・エー/エス 安定化ポリペプチド製剤
US8097584B2 (en) * 2005-05-25 2012-01-17 Novo Nordisk A/S Stabilized formulations of insulin that comprise ethylenediamine
WO2007121318A2 (fr) 2006-04-12 2007-10-25 Emisphere Technologies, Inc. Préparations destinées à l'administration d'insuline
EP2040718B1 (fr) 2006-06-28 2017-12-27 Emisphere Technologies, Inc. Formulations de nitrate de gallium
WO2008132731A2 (fr) * 2007-04-26 2008-11-06 Oramed Pharmaceuticals, Inc Procédés et compositions pour administration rectale d'insuline
IL188647A0 (en) * 2008-01-08 2008-11-03 Orina Gribova Adaptable structured drug and supplements administration system (for oral and/or transdermal applications)
AU2009283821B2 (en) 2008-08-18 2014-05-29 Entera Bio Ltd. Methods and compositions for oral administration of proteins
WO2011004376A1 (fr) 2009-07-09 2011-01-13 Oshadi Drug Administration Ltd. Compositions de support matriciel, procédés et utilisations
WO2023286049A1 (fr) 2021-07-12 2023-01-19 Zidkiyahu Simenhaus Compositions bioactives contenant des protéines et comprenant des supports de microparticules de cellulose

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4863896A (en) 1984-05-03 1989-09-05 Technology Unlimited, Inc. Diabetic control by combined insulin forms
RU2066551C1 (ru) 1994-03-23 1996-09-20 Институт нефтехимического синтеза РАН Способ получения инсулинсодержащих полимерных гидрогелей
US5780243A (en) * 1996-06-28 1998-07-14 Thaco Research, Ltd. Methods for the quantitative analysis of organic compounds
SE520392C2 (sv) * 1996-09-27 2003-07-01 Creative Peptides Sweden Ab C Specifika peptider för behandling av diabetes mellitus
US6197340B1 (en) * 1998-05-28 2001-03-06 Medical Research Institute Controlled release lipoic acid
AU2001273153B2 (en) 2000-06-29 2005-11-17 Emisphere Technologies, Inc. Compounds and compositions for delivering active agents
US7115663B2 (en) * 2000-09-06 2006-10-03 Emisphere Technologies, Inc. Cyanophenoxy carboxylic acid compounds and compositions for delivering active agents
US20020147135A1 (en) * 2000-12-21 2002-10-10 Oliver Schnell Method and device for producing an adapted travel treatment plan for administering a medicine in the event of a long-haul journey
US7060675B2 (en) * 2001-02-15 2006-06-13 Nobex Corporation Methods of treating diabetes mellitus
US6828297B2 (en) * 2001-06-04 2004-12-07 Nobex Corporation Mixtures of insulin drug-oligomer conjugates comprising polyalkylene glycol, uses thereof, and methods of making same
US20030198666A1 (en) * 2002-01-07 2003-10-23 Richat Abbas Oral insulin therapy
EP1469827B1 (fr) 2002-01-09 2017-12-27 Emisphere Technologies, Inc. Polymorphes de 4- (4-chloro-2-hydroxybenzoyl)amino butanoate de sodium
US7137951B2 (en) * 2002-10-23 2006-11-21 Joseph Pilarski Method of food and insulin dose management for a diabetic subject
CA2565188C (fr) 2004-05-14 2014-03-04 Emisphere Technologies, Inc. Composes et compositions pour administration d'agents actifs

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
None
See also references of EP1592438A4

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8492330B2 (en) 2002-02-20 2013-07-23 Emisphere Technologies, Inc. Formulation comprising GLP-1
US7939494B2 (en) 2002-02-20 2011-05-10 Emisphere Technologies, Inc. Method for administering GLP-1 molecules
US8324156B2 (en) 2003-08-22 2012-12-04 Emisphere Technologies, Inc. Oral insulin therapies and protocol
US8729016B2 (en) 2003-08-22 2014-05-20 Emisphere Technologies, Inc. Oral insulin therapies and protocol
US8962554B2 (en) 2003-08-22 2015-02-24 Emisphere Technologies, Inc. Oral insulin therapies and protocol
US8785381B2 (en) 2003-12-19 2014-07-22 Emisphere Technologies, Inc. Oral GLP-1 formulations
EP2279732A2 (fr) 2004-05-14 2011-02-02 Emisphere Technologies, Inc. Composés et compositions pour la distribution d'agents actifs
WO2005112937A1 (fr) 2004-05-19 2005-12-01 Emisphere Technologies, Inc. Formulations d’acyclovir
EP2248531A1 (fr) * 2004-08-03 2010-11-10 Emisphere Technologies, Inc. Combinaison orale antidiabétique d'insuline-biguanide
JP2008509145A (ja) * 2004-08-03 2008-03-27 エミスフィアー テクノロジーズ インコーポレイテッド 抗糖尿病性経口インスリン−ビグアニドの組み合わせ
WO2006072070A2 (fr) 2004-12-29 2006-07-06 Emisphere Technologies, Inc. Formulations pharmaceutiques contenant des sels de gallium
US8110547B2 (en) 2005-01-12 2012-02-07 Emisphere Technologies, Inc. Compositions for buccal delivery of parathyroid hormone
US8771712B2 (en) 2006-05-09 2014-07-08 Emisphere Technologies, Inc. Topical administration of acyclovir
WO2011017346A2 (fr) 2009-08-03 2011-02-10 Emisphere Technologies, Inc. Composition de naproxène à action rapide avec effets gastro-intestinaux réduits

Also Published As

Publication number Publication date
WO2004062587A3 (fr) 2005-04-28
EP1592438A4 (fr) 2009-07-08
ES2465496T3 (es) 2014-06-05
EP1592438A2 (fr) 2005-11-09
US20060178296A1 (en) 2006-08-10
NZ541058A (en) 2008-06-30
AU2004204727B8 (en) 2006-11-09
AU2004204727A1 (en) 2004-07-29
JP2006515620A (ja) 2006-06-01
HK1085133A1 (en) 2006-08-18
CA2511530A1 (fr) 2004-07-29
JP5452843B2 (ja) 2014-03-26
AU2004204727B2 (en) 2006-10-19
CA2511530C (fr) 2013-07-09
US7384914B2 (en) 2008-06-10
EP1592438B1 (fr) 2014-02-26

Similar Documents

Publication Publication Date Title
US7384914B2 (en) Night-time oral insulin therapy
US7429564B2 (en) Oral insulin therapy
US20180161400A1 (en) Antidiabetic oral insulin biguanide combination
US8962554B2 (en) Oral insulin therapies and protocol
AU2004220544B2 (en) Oral insulin therapies and protocol
ZA200505426B (en) Night-time oral insulin therapy

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2511530

Country of ref document: CA

Ref document number: 2004204727

Country of ref document: AU

Ref document number: 541058

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2005/05426

Country of ref document: ZA

Ref document number: 2994/DELNP/2005

Country of ref document: IN

Ref document number: 200505426

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 2006500813

Country of ref document: JP

DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)
WWP Wipo information: published in national office

Ref document number: 2004204727

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2004700388

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 20048052617

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2004700388

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2006178296

Country of ref document: US

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 10541433

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 10541433

Country of ref document: US